1
|
Liu C, Zhao K, Chen Y, Yao Y, Tang J, Wang J, Xu C, Yang Q, Zheng Y, Yuan Y, Sun H, Zhang Y, Zhou Y, Chen J, Wang Y, Wu C, Pei R, Chen X. Mitochondrial Glycerol-3-Phosphate Dehydrogenase Restricts HBV Replication via the TRIM28-Mediated Degradation of HBx. J Virol 2023; 97:e0058023. [PMID: 37166302 PMCID: PMC10231258 DOI: 10.1128/jvi.00580-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/12/2023] Open
Abstract
Hepatitis B virus (HBV) infection affects hepatic metabolism. Serum metabolomics studies have suggested that HBV possibly hijacks the glycerol-3-phosphate (G3P) shuttle. In this study, the two glycerol-3-phosphate dehydrogenases (GPD1 and GPD2) in the G3P shuttle were analyzed for determining their role in HBV replication and the findings revealed that GPD2 and not GPD1 inhibited HBV replication. The knockdown of GPD2 expression upregulated HBV replication, while GPD2 overexpression reduced HBV replication. Moreover, the overexpression of GPD2 significantly reduced HBV replication in hydrodynamic injection-based mouse models. Mechanistically, this inhibitory effect is related to the GPD2-mediated degradation of HBx protein by recruiting the E3 ubiquitin ligase TRIM28 and not to the alterations in G3P metabolism. In conclusion, this study revealed GPD2, a key enzyme in the G3P shuttle, as a host restriction factor in HBV replication. IMPORTANCE The glycerol-3-phosphate (G3P) shuttle is important for the delivery of cytosolic reducing equivalents into mitochondria for oxidative phosphorylation. The study analyzed two key components of the G3P shuttle and identified GPD2 as a restriction factor in HBV replication. The findings revealed a novel mechanism of GPD2-mediated inhibition of HBV replication via the recruitment of TRIM28 for degrading HBx, and the HBx-GPD2 interaction could be another potential therapeutic target for anti-HBV drug development.
Collapse
Affiliation(s)
- Canyu Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kaitao Zhao
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yingshan Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongxuan Yao
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Jielin Tang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- Guangzhou Laboratory, Guangzhou, China
| | - Jingjing Wang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Chonghui Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qi Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- Guangzhou Laboratory, Guangzhou, China
| | - Yi Zheng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yifei Yuan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongli Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
| | - Yuan Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
| | | | - Yun Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
| | - Chunchen Wu
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rongjuan Pei
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
| | - Xinwen Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei Province, China
- Guangzhou Laboratory, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
40 Years of Research on Polybrominated Diphenyl Ethers (PBDEs)-A Historical Overview and Newest Data of a Promising Anticancer Drug. Molecules 2021; 26:molecules26040995. [PMID: 33668501 PMCID: PMC7918430 DOI: 10.3390/molecules26040995] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are a group of molecules with an ambiguous background in literature. PBDEs were first isolated from marine sponges of Dysidea species in 1981 and have been under continuous research to the present day. This article summarizes the two research aspects, (i) the marine compound chemistry research dealing with naturally produced PBDEs and (ii) the environmental toxicology research dealing with synthetically-produced brominated flame-retardant PBDEs. The different bioactivity patterns are set in relation to the structural similarities and dissimilarities between both groups. In addition, this article gives a first structure-activity relationship analysis comparing both groups of PBDEs. Moreover, we provide novel data of a promising anticancer therapeutic PBDE (i.e., 4,5,6-tribromo-2-(2',4'-dibromophenoxy)phenol; termed P01F08). It has been known since 1995 that P01F08 exhibits anticancer activity, but the detailed mechanism remains poorly understood. Only recently, Mayer and colleagues identified a therapeutic window for P01F08, specifically targeting primary malignant cells in a low µM range. To elucidate the mechanistic pathway of cell death induction, we verified and compared its cytotoxicity and apoptosis induction capacity in Ramos and Jurkat lymphoma cells. Moreover, using Jurkat cells overexpressing antiapoptotic Bcl-2, we were able to show that P01F08 induces apoptosis mainly through the intrinsic mitochondrial pathway.
Collapse
|
3
|
Shi X, Zha J, Wen B, Zhang S. Diastereoisomer-specific neurotoxicity of hexabromocyclododecane in human SH-SY5Y neuroblastoma cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 686:893-902. [PMID: 31200309 DOI: 10.1016/j.scitotenv.2019.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/29/2019] [Accepted: 06/01/2019] [Indexed: 06/09/2023]
Abstract
Hexabromocyclododecane (HBCD) is a widely applied brominated flame retardant (BFR) and is regarded as a persistent organic pollutant. It has been found in human tissues and has the potential to cause neurological disorders. However, our understanding of HBCD neurotoxicity at the diastereoisomer level remains lacking. Here, we investigated the neurotoxicity of three HBCD diastereoisomers, i.e., α-, β-, and γ-HBCD, in SH-SY5Y human neuroblastoma cells. Results showed that the HBCD diastereoisomers decreased cell viability, increased lactate dehydrogenase (LDH) release, and impaired cytoskeleton development. Typical morphological features and apoptosis rates showed that the HBCD diastereoisomers induced SH-SY5Y cell apoptosis. The expression levels of several cell apoptosis-related genes and proteins, including Bax, caspase-3, caspase-9, cytochrome c, Bcl-2, and X-linked inhibitor of apoptosis (XIAP), as well as the cell cycle arrest, DNA damage, adenosine triphosphate (ATP) consumption, reactive oxygen species (ROS) levels, and intracellular calcium ion (Ca2+) levels, were examined. Results showed that the HBCD diastereoisomer neurotoxicity was ranked β-HBCD > γ-HBCD > α-HBCD. The cell apoptosis and caspase expression levels of the three HBCD diastereoisomers followed the same order, suggesting that caspase-dependent apoptosis may be one mechanism responsible for the structure-selective HBCD diastereoisomer neurotoxicity. The levels of intracellular Ca2+ and ROS increased significantly. The ROS levels were ordered β-HBCD > γ-HBCD > α-HBCD, whereas those of intracellular Ca2+ were γ-HBCD > β-HBCD > α-HBCD. Thus, ROS may be a key factor regulating the neurotoxicity of HBCD diastereoisomers. To the best of our knowledge, this is the first study to report on the diastereoisomer-specific toxicity of HBCD in human neural cells and on the possible mechanisms responsible for the selective neurotoxicity of HBCD diastereoisomers.
Collapse
Affiliation(s)
- Xiaoli Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinmiao Zha
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bei Wen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Shuzhen Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
4
|
Li M, Huo X, Pan Y, Cai H, Dai Y, Xu X. Proteomic evaluation of human umbilical cord tissue exposed to polybrominated diphenyl ethers in an e-waste recycling area. ENVIRONMENT INTERNATIONAL 2018; 111:362-371. [PMID: 29169793 DOI: 10.1016/j.envint.2017.09.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/17/2017] [Accepted: 09/19/2017] [Indexed: 02/05/2023]
Abstract
Parental exposure to polybrominated diphenyl ethers (PBDEs) is associated with adverse birth outcomes. This study aims to examine differentially-expressed protein profiles in umbilical cord tissue, derived from mothers exposed to PBDEs, and investigate candidate biomarkers to reveal the underlying molecular mechanisms. Umbilical cord samples were obtained from women residing in an electronic waste (e-waste) recycling area (Guiyu) and reference area (Haojiang) in China. The concentration of PBDEs in umbilical cord tissue was determined by gas chromatography and mass spectrometry (GC/MS). Isobaric tagging for relative and absolute quantification (iTRAQ)-based proteomic technology was conducted to analyze differentially-expressed protein profiles. The total PBDE concentration was approximately five-fold higher in umbilical cords from Guiyu than from Haojiang (median 71.92ng/g vs. 15.52ng/g lipid, P<0.01). Neonatal head circumference, body-mass index (BMI) and Apgar1 score were lower in Guiyu and negatively correlated with PBDE concentration (P<0.01). Proteomic analysis showed 697 proteins were differentially expressed in the e-waste-exposed group compared with the reference group. The differentially-expressed proteins were principally involved in antioxidant defense, apoptosis, cell structure and metabolism. Among them, catalase and glutathione S-transferase omega-1, were down-regulated, and cytochrome c was found to be up-regulated, changes which were further verified by enzyme-linked immunosorbent assays. These results suggest that an antioxidant imbalance and cell apoptosis in the umbilical cord following PBDE exposure is associated with neonatal birth outcomes.
Collapse
Affiliation(s)
- Minghui Li
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangzhou Key Laboratory of Environmental Exposure and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Yukui Pan
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Haoxing Cai
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yifeng Dai
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
5
|
Wong S, Giulivi C. Autism, Mitochondria and Polybrominated Diphenyl Ether Exposure. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2017; 15:614-23. [PMID: 27071785 DOI: 10.2174/1871527315666160413122624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/29/2015] [Accepted: 01/09/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Autism spectrum disorders (ASD) are a growing concern with more than 1 in every 68 children affected in the United States by age 8. Limited scientific advances have been made regarding the etiology of autism, with general agreement that both genetic and environmental factors contribute to this disorder. OBJECTIVE To explore the link between exposure to PBDE, mitochondrial dysfunction and autism risk. RESULTS Perinatal exposures to PBDEs may contribute to the etiology or morbidity of ASD including mitochondrial dysfunction based on (i) their increased environmental abundance and human exposures, (ii) their activity towards implicated in neuronal development and synaptic plasticity including mitochondria, and (iii) their bioaccumulation in mitochondria. CONCLUSION In this review, we propose that PBDE, and possibly other environmental exposures, during child development can induce or compound mitochondrial dysfunction, which in conjunction with a dysregulated antioxidant response, increase a child's susceptibility of autism.
Collapse
Affiliation(s)
| | - Cecilia Giulivi
- University of California, Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., 3009 VetMed3B, Davis, CA 95616, USA.
| |
Collapse
|
6
|
Fournier K, Baumont E, Glorennec P, Bonvallot N. Relative toxicity for indoor semi volatile organic compounds based on neuronal death. Toxicol Lett 2017; 279:33-42. [PMID: 28709981 DOI: 10.1016/j.toxlet.2017.07.875] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 07/03/2017] [Accepted: 07/09/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND Semi Volatile Organic Compounds (SVOCs) are contaminants commonly found in dwellings as a result of their use as plasticizers, flame retardants, or pesticides in building materials and consumer products. Many SVOCs are suspected of being neurotoxic, based on mammal experimentation (impairment of locomotor activity, spatial learning/memory or behavioral changes), raising the question of cumulative risk assessment. The aim of this work is to estimate the relative toxicity of such SVOCs, based on neuronal death. METHOD SVOCs fulfilling the following conditions were included: detection frequency >10% in dwellings, availability of data on effects or mechanism of action for neurotoxicity, and availability of dose-response relationships based on cell viability assays as a proxy of neuronal death. Benchmark concentration values (BMC) were estimated using a Hill model, and compared to assess relative toxicity. RESULTS Of the 58 SVOCs selected, 28 were suspected of being neurotoxic in mammals, and 21 have been documented as inducing a decrease in cell viability in vitro. 13 have at least one dose-response relationship that can be used to derive a BMC based on a 10% fall in neuronal viability. Based on this in vitro endpoint, PCB-153 appeared to be the most toxic compound, having the lowest BMC10 (0.072μM) and diazinon the least toxic compound, having the highest BMC10 (94.35μM). We showed that experimental designs (in particular choice of cell lines) had a significant influence on BMC calculation. CONCLUSION For the first time, the relative in vitro toxicity of 13 indoor contaminants belonging to different chemical families has been assessed on the basis of neuronal cell viability. Lack of comparable toxicity datasets limits the number of SVOCs that can be included. More standardized protocols in terms of cell lines, species and exposure duration should be developed with a view to cumulative risk assessment.
Collapse
Affiliation(s)
- Kevin Fournier
- EHESP School of Public Health, Sorbonne Paris Cité, Avenue du Professeur Léon Bernard, 35043 Rennes Cedex, France; INSERM UMR1085 IRSET (Research Institute in Environmental and Occupational Health), Rennes, France.
| | - Emmanuel Baumont
- EHESP School of Public Health, Sorbonne Paris Cité, Avenue du Professeur Léon Bernard, 35043 Rennes Cedex, France; INSERM UMR1085 IRSET (Research Institute in Environmental and Occupational Health), Rennes, France.
| | - Philippe Glorennec
- EHESP School of Public Health, Sorbonne Paris Cité, Avenue du Professeur Léon Bernard, 35043 Rennes Cedex, France; INSERM UMR1085 IRSET (Research Institute in Environmental and Occupational Health), Rennes, France.
| | - Nathalie Bonvallot
- EHESP School of Public Health, Sorbonne Paris Cité, Avenue du Professeur Léon Bernard, 35043 Rennes Cedex, France; INSERM UMR1085 IRSET (Research Institute in Environmental and Occupational Health), Rennes, France.
| |
Collapse
|
7
|
Sharma N, Baek K, Phan HTT, Shimokawa N, Takagi M. Glycosyl chains and 25-hydroxycholesterol contribute to the intracellular transport of amyloid beta (Aβ-42) in Jurkat T cells. FEBS Open Bio 2017; 7:865-876. [PMID: 28593141 PMCID: PMC5458452 DOI: 10.1002/2211-5463.12234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 01/05/2023] Open
Abstract
Amyloid beta (Aβ) is a peptide responsible for the development of Alzheimer's disease (AD). Misfolding and accumulation of endogenous Aβ can lead to neural cell apoptosis through endoplasmic reticulum (ER) stress. Added exogenous Aβ can also result in ER stress, leading to neurotoxicity and apoptosis, which is identical to that caused by the endogenous peptide. We have speculated that the endocytic transport of Aβ causes ER stress and have previously shown that the oxysterol, in particular, 7-ketocholesterol (7-keto) induces more surface interaction between Aβ-42 and Jurkat cells than cholesterol. However, the interaction was not enough to induce intracellular transfer of the peptide. In this study, we investigated the effect of another oxysterol, 25-hydroxycholesterol (25-OH) on the membrane raft-dependent transport of Aβ-42 in Jurkat cells. Interestingly, intracellular transfer of Aβ-42 was observed in the presence of 25-OH only after the inclusion of cholera toxin B subunit (CT-B), a marker used to detect the raft domain. We speculated that 25-OH can induce intracellular movement of Aβ peptides. Furthermore, CT-B together with GM1 provided negative curvature, which resulted in the intracellular transport of Aβ-42. Notably, we used a protofibrillar species of Aβ-42 in this study. We have shown that the transport was microtubule-dependent since it could not be observed in depolymerized microtubules. These results demonstrate that oxysterols and glycosyl chains are important factors affecting intracellular transport. These compounds are also associated with aging and advanced glycation are risk factors for AD. Thus, this study should further understanding of the pathology of AD.
Collapse
Affiliation(s)
- Neha Sharma
- School of Materials Science Japan Advanced Institute of Science and Technology (JAIST) Ishikawa Japan
| | - KeangOK Baek
- School of Materials Science Japan Advanced Institute of Science and Technology (JAIST) Ishikawa Japan
| | | | - Naofumi Shimokawa
- School of Materials Science Japan Advanced Institute of Science and Technology (JAIST) Ishikawa Japan
| | - Masahiro Takagi
- School of Materials Science Japan Advanced Institute of Science and Technology (JAIST) Ishikawa Japan
| |
Collapse
|
8
|
Dong Z, Hu Z, Zhu H, Li N, Zhao H, Mi W, Jiang W, Hu X, Ye L. Tris-(2,3-dibromopropyl) isocyanurate induces depression-like behaviors and neurotoxicity by oxidative damage and cell apoptosis in vitro and in vivo. J Toxicol Sci 2016; 40:701-9. [PMID: 26558450 DOI: 10.2131/jts.40.701] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Tris-(2,3-dibromopropyl) isocyanurate (TDBP-TAZTO), an emerging brominated flame retardant, possesses the characteristics of candidate persistent organic pollutants and has displayed toxicity to fish and rodents. TDBP-TAZTO can pass through the blood-brain barrier and accumulate in the brain. TDBP-TAZTO might also induce neuronal cell toxicity. However, the neurotoxicity and mechanisms of TDBP-TAZTO have not yet been studied. We hypothesize that TDBP-TAZTO could induce neurotoxicity in mouse hippocampal neurons and SH-SY5Y cells. The mice were exposed to TDBP-TAZTO of 5 and 50 mg/kg by gavage, daily for 30 days. TDBP-TAZTO resulted in depression-like behaviors, which may be related with TDBP-TAZTO-induced upregulation of oxidative stress markers and overexpression of pro-apoptotic proteins in hippocampus. Furthermore, TDBP-TAZTO treatment for 48 hr (12.5, 25 and 50 µM) damaged SH-SY5Y cells, and led to cell apoptosis and oxidative stress in concentration-dependent manner. Our findings suggested that cell apoptosis and oxidative stress are important mechanisms in neurotoxicity induced by TDBP-TAZTO.
Collapse
Affiliation(s)
- Zhaoju Dong
- School of Public Health and Management, Binzhou Medical University, China
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Chen H, Tang X, Zhou B, Xu N, Wang Y. Mechanism of Deca-BDE-induced apoptosis in Neuro-2a cells: Role of death-receptor pathway and reactive oxygen species-mediated mitochondrial pathway. J Environ Sci (China) 2016; 46:241-251. [PMID: 27521956 DOI: 10.1016/j.jes.2016.02.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/31/2016] [Accepted: 02/04/2016] [Indexed: 06/06/2023]
Affiliation(s)
- Hongmei Chen
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Xuexi Tang
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Bin Zhou
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Ningning Xu
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - You Wang
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
10
|
Lefevre PLC, Wade M, Goodyer C, Hales BF, Robaire B. A Mixture Reflecting Polybrominated Diphenyl Ether (PBDE) Profiles Detected in Human Follicular Fluid Significantly Affects Steroidogenesis and Induces Oxidative Stress in a Female Human Granulosa Cell Line. Endocrinology 2016; 157:2698-711. [PMID: 27219277 DOI: 10.1210/en.2016-1106] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Brominated flame retardants are incorporated into consumer products to prevent flame propagation. These compounds leach into the domestic environment, resulting in chronic exposure. Pregnancy failure is associated with high levels of polybrominated diphenyl ethers (PBDEs), a major class of brominated flame retardants, in human follicular fluid, raising serious questions regarding their impact on female fertility. Our goal was to elucidate the effects of a mixture of PBDEs, similar to the profile found in human follicular fluid, on an immortalized human granulosa cell line, the KGN cell line. We showed that cell viability was altered and oxidative stress was induced as reflected by increased reactive oxygen species formation at 100 μM of the PBDE mixture. Transcriptomic analysis revealed that PBDE treatments of 1, 5, and 20 μM altered the expression of several genes involved in the reactive oxygen species signaling pathway. Significant dose-dependent reductions in progesterone and estradiol levels in the culture medium were measured after PBDE treatment; in parallel, the expression of genes involved in estradiol metabolism, namely CYP1A1, was up-regulated by 5 and 20 μM of the PBDE mixture. Treatment with 20 μM PBDE also increased the expression and secretion of the proinflammatory factor, IL-6, into the KGN cell culture medium. Our results demonstrate that PBDEs can alter human granulosa cell functions by inducing oxidative stress and disrupting steroidogenesis. These results indicate that PBDEs may be detrimental to ovarian functions and thus may adversely affect female reproductive health after chronic exposure.
Collapse
Affiliation(s)
- Pavine L C Lefevre
- Departments of Pharmacology and Therapeutics (P.L.C.L., B.F.H., B.R.), Pediatrics (C.G.), and Obstetrics and Gynecology (B.R.), McGill University, and the Research Institute of the McGill University Health Centre, Montréal, Québec, Canada H3G 1Y6; and Environmental Health Science and Research Bureau (M.W.), Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Mike Wade
- Departments of Pharmacology and Therapeutics (P.L.C.L., B.F.H., B.R.), Pediatrics (C.G.), and Obstetrics and Gynecology (B.R.), McGill University, and the Research Institute of the McGill University Health Centre, Montréal, Québec, Canada H3G 1Y6; and Environmental Health Science and Research Bureau (M.W.), Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Cindy Goodyer
- Departments of Pharmacology and Therapeutics (P.L.C.L., B.F.H., B.R.), Pediatrics (C.G.), and Obstetrics and Gynecology (B.R.), McGill University, and the Research Institute of the McGill University Health Centre, Montréal, Québec, Canada H3G 1Y6; and Environmental Health Science and Research Bureau (M.W.), Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Barbara F Hales
- Departments of Pharmacology and Therapeutics (P.L.C.L., B.F.H., B.R.), Pediatrics (C.G.), and Obstetrics and Gynecology (B.R.), McGill University, and the Research Institute of the McGill University Health Centre, Montréal, Québec, Canada H3G 1Y6; and Environmental Health Science and Research Bureau (M.W.), Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Bernard Robaire
- Departments of Pharmacology and Therapeutics (P.L.C.L., B.F.H., B.R.), Pediatrics (C.G.), and Obstetrics and Gynecology (B.R.), McGill University, and the Research Institute of the McGill University Health Centre, Montréal, Québec, Canada H3G 1Y6; and Environmental Health Science and Research Bureau (M.W.), Health Canada, Ottawa, Ontario, Canada K1A 0K9
| |
Collapse
|
11
|
Souza AO, Tasso MJ, Oliveira AMC, Pereira LC, Duarte FV, Oliveira DP, Palmeira CM, Dorta DJ. Evaluation of Polybrominated Diphenyl Ether Toxicity on HepG2 Cells - Hexabrominated Congener (BDE-154) Is Less Toxic than Tetrabrominated Congener (BDE-47). Basic Clin Pharmacol Toxicol 2016; 119:485-497. [DOI: 10.1111/bcpt.12598] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/04/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Alecsandra O. Souza
- Department of Chemistry; Faculty of Philosophy; Sciences and Languages of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP Brazil
| | - Maria J. Tasso
- Department of Chemistry; Faculty of Philosophy; Sciences and Languages of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP Brazil
| | - Alana M. C. Oliveira
- Department of Chemistry; Faculty of Philosophy; Sciences and Languages of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP Brazil
| | - Lilian C. Pereira
- Department of Clinical Analysis, Toxicological and Bromatological; Faculty of Pharmaceutical Sciences of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP Brazil
| | - Filipe V. Duarte
- CNC - Center for Neuroscience and Cell Biology; Faculty of Medicine; University of Coimbra; Coimbra Portugal
- Department of Life Sciences; University of Coimbra; Coimbra Portugal
| | - Danielle P. Oliveira
- Department of Clinical Analysis, Toxicological and Bromatological; Faculty of Pharmaceutical Sciences of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP Brazil
| | - Carlos M. Palmeira
- CNC - Center for Neuroscience and Cell Biology; Faculty of Medicine; University of Coimbra; Coimbra Portugal
- Department of Life Sciences; University of Coimbra; Coimbra Portugal
| | - Daniel J. Dorta
- Department of Chemistry; Faculty of Philosophy; Sciences and Languages of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP Brazil
| |
Collapse
|
12
|
Pentoxifylline triggers autophagy via ER stress response that interferes with Pentoxifylline induced apoptosis in human melanoma cells. Biochem Pharmacol 2016; 103:17-28. [PMID: 26793997 DOI: 10.1016/j.bcp.2015.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 12/22/2015] [Indexed: 12/17/2022]
Abstract
Pentoxifylline (PTX), a non-specific phosphodiesterase inhibitor is known to inhibit the growth of various cancer cells including melanoma. Here in this study, we have found that PTX induces autophagy in human melanoma cell lines (A375 and MeWo). Induction of autophagy is associated with the increase in Atg5 expression as knockdown of Atg5 effectively inhibited PTX mediated autophagy. A decrease in mTOR activation was also observed after PTX treatment. We observed that autophagy was activated as a downstream effector mechanism of ER stress induced by PTX. ER stress response was confirmed by upregulation of IRE-1α, GRP78 and CHOP expression. PTX treatment also resulted in an increase in intracellular calcium (Ca(2+)) level. Ca(2+) is the central player as blocking Ca(2+) by intracellular calcium chelator (BAPTA-AM) effectively inhibited the PTX induced ER stress response as well as autophagy. Moreover, silencing of CHOP also resulted in autophagy inhibition with a decrease in Atg5 expression. Collectively, PTX triggers ER stress response followed by induction of autophagy via involvement of Ca(2+)→CHOP→Atg5 signalling cascade. Interestingly, inhibition of intracellular calcium level by BAPTA-AM significantly increased PTX mediated cell death by augmenting intrinsic apoptotic pathway. Inhibition of autophagy by the ATG5 siRNA and pharmacological inhibitor, chloroquine also enhances PTX induced cell death. Taken together, our results clearly indicate that activation of ER stress response and autophagy provides resistance to PTX mediated apoptosis, and thus, interferes with the anticancer activity of PTX in human melanoma cells.
Collapse
|
13
|
The Pivotal Role of Ca 2+ Homeostasis in PBDE-47-Induced Neuronal Apoptosis. Mol Neurobiol 2015; 53:7078-7088. [PMID: 26676572 DOI: 10.1007/s12035-015-9573-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/29/2015] [Indexed: 10/22/2022]
Abstract
Polybrominated diphenyl ethers (PBDEs) are widely used flame retardants and are ubiquitous in the environment and human tissues. Recent evidence has demonstrated that PBDE-induced neurotoxicity is associated with neuronal apoptosis via interfering with the calcium ion (Ca2+) homeostasis; however, the underlying mechanisms remain elusive. Thus, we sought to investigate the role of Ca2+ homeostasis in PBDE-47-induced neuronal apoptosis. Here, we showed that PBDE-47 significantly decreased neuronal number while increased neuronal apoptosis in vitro and in vivo, as manifested by an increased percentage of Annexin V-positive staining cells and caspase-3 activation in human neuroblastoma SH-SY5Y cells and hippocampal neurons of rats. Further study identified that PBDE-47 elicited ΔΨm collapse following an early and sustained [Ca2+] i, overload, as well as stimulated cytochrome c release from mitochondria into the cytosol in SH-SY5Y cells and rat hippocampal tissue. Interestingly, the extracellular Ca2+ chelator ethylene glycol-bis (2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA) blocked PBDE-47-induced [Ca2+] i elevation, ΔΨm collapse, cytochrome c release, and caspase-3 activation in SH-SY5Y cells, whereas the intracellular Ca2+ chelator 1,2-bis (2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (BAPTA/AM) had no influences on them, indicating that the [Ca2+] i overload originates primarily from extracellular Ca2+ component rather than from intracellular calcium storage and that the increase in [Ca2+] i is a major contributor to ΔΨm collapse and subsequent neuronal apoptosis. Overall, these findings suggest that PBDE-47 affects Ca2+ homeostasis as a crucial event in activation of neuronal death associated with mitochondria and provide novel insight into the mechanism of action underlying PBDE neurotoxicity.
Collapse
|
14
|
Role of glutamate receptors in tetrabrominated diphenyl ether (BDE-47) neurotoxicity in mouse cerebellar granule neurons. Toxicol Lett 2015; 241:159-66. [PMID: 26640238 DOI: 10.1016/j.toxlet.2015.11.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/25/2015] [Accepted: 11/25/2015] [Indexed: 01/22/2023]
Abstract
The polybrominated diphenyl ether (PBDE) flame retardants are developmental neurotoxicants, as evidenced by numerous in vitro, animal and human studies. PBDEs can alter the homeostasis of thyroid hormone and directly interact with brain cells. Induction of oxidative stress, leading to DNA damage and apoptotic cell death is a prominent mechanism of PBDE neurotoxicity, though other mechanisms have also been suggested. In the present study we investigated the potential role played by glutamate receptors in the in vitro neurotoxicity of the tetrabromodiphenyl ether BDE-47, one of the most abundant PBDE congeners. Toxicity of BDE-47 in mouse cerebellar neurons was diminished by antagonists of glutamate ionotropic receptors, but not by antagonists of glutamate metabotropic receptors. Antagonists of NMDA and AMPA/Kainate receptors also inhibited BDE-47-induced oxidative stress and increases in intracellular calcium. The calcium chelator BAPTA-AM also inhibited BDE-47 cytotoxicity and oxidative stress. BDE-47 caused a rapid increase of extracellular glutamate levels, which was not antagonized by any of the compounds tested. The results suggest that BDE-47, by still unknown mechanisms, increases extracellular glutamate which in turn activates ionotropic glutamate receptors leading to increased calcium levels, oxidative stress, and ultimately cell death.
Collapse
|
15
|
Hendriks HS, Westerink RH. Neurotoxicity and risk assessment of brominated and alternative flame retardants. Neurotoxicol Teratol 2015; 52:248-69. [DOI: 10.1016/j.ntt.2015.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 09/01/2015] [Accepted: 09/01/2015] [Indexed: 11/29/2022]
|
16
|
Wang X, Yang L, Wu Y, Huang C, Wang Q, Han J, Guo Y, Shi X, Zhou B. The developmental neurotoxicity of polybrominated diphenyl ethers: Effect of DE-71 on dopamine in zebrafish larvae. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2015; 34:1119-1126. [PMID: 25651517 DOI: 10.1002/etc.2906] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/27/2014] [Accepted: 01/21/2015] [Indexed: 06/04/2023]
Abstract
The potential neurotoxicity of polybrominated diphenyl ethers (PBDEs) is still a great concern. In the present study, the authors investigated whether exposure to PBDEs could affect the neurotransmitter system and cause developmental neurotoxicity in zebrafish. Zebrafish embryos (2 h postfertilization) were exposed to different concentrations of the PBDE mixture DE-71 (0-100 μg/L). The larvae were harvested at 120 h postfertilization, and the impact on dopaminergic signaling was investigated. The results revealed significant reductions in content of whole-body dopamine and its metabolite, dihydroxyphenylacetic acid, in DE-71-exposed larvae. The transcription of genes involved in the development of dopaminergic neurons (e.g., manf, bdnf, and nr4a2b) was significantly downregulated upon exposure to DE-71. Also, DE-71 resulted in a significant decrease of tyrosine hydroxylase and dopamine transporter protein levels in dopaminergic neurons. The expression level of tyrosine hydroxylase in forebrain neurons was assessed by whole-mount immunofluorescence, and the results further demonstrated that the tyrosine hydroxylase protein expression level was reduced in dopaminergic neurons. In addition to these molecular changes, the authors observed reduced locomotor activity in DE-71-exposed larvae. Taken together, the results of the present study demonstrate that acute exposure to PBDEs can affect dopaminergic signaling by disrupting the synthesis and transportation of dopamine in zebrafish, thereby disrupting normal neurodevelopment. In accord with its experimental findings, the present study extends knowledge of the mechanisms governing PBDE-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Xianfeng Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sulaiman MK, Chu Z, Blanco VM, Vallabhapurapu SD, Franco RS, Qi X. SapC-DOPS nanovesicles induce Smac- and Bax-dependent apoptosis through mitochondrial activation in neuroblastomas. Mol Cancer 2015; 14:78. [PMID: 25889084 PMCID: PMC4397704 DOI: 10.1186/s12943-015-0336-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 03/09/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND High toxicity, morbidity and secondary malignancy render chemotherapy of neuroblastoma inefficient, prompting the search for novel compounds. Nanovesicles offer great promise in imaging and treatment of cancer. SapC-DOPS, a stable nanovesicle formed from the lysosomal protein saposin C and dioleoylphosphatidylserine possess strong affinity for abundantly exposed surface phosphatidylserine on cancer cells. Here, we show that SapC-DOPS effectively targets and suppresses neuroblastoma growth and elucidate the molecular mechanism of SapC-DOPS action in neuroblastoma in vitro. METHODS In vivo targeting of neuroblastoma was assessed in xenograft mice injected intravenously with fluorescently-labeled SapC-DOPS. Xenografted tumors were also used to demonstrate its therapeutic efficacy. Apoptosis induction in vivo was evaluated in tumor sections using the TUNEL assay. The mechanisms underlying the induction of apoptosis by SapC-DOPS were addressed through measurements of cell viability, mitochondrial membrane potential (ΔΨM), flow cytometric DNA fragmentation assays and by immunoblot analysis of second mitochondria-derived activator of caspases (Smac), Bax, Cytochrome c (Cyto c) and Caspase-3 in the cytosol or in mitochondrial fractions of cultured neuroblastoma cells. RESULTS SapC-DOPS showed specific targeting and prevented the growth of human neuroblastoma xenografts in mice. In neuroblastoma cells in vitro, apoptosis occurred via a series of steps that included: (1) loss of ΔΨM and increased mitochondrial superoxide formation; (2) cytosolic release of Smac, Cyto c, AIF; and (3) mitochondrial translocation and polymerization of Bax. ShRNA-mediated Smac knockdown and V5 peptide-mediated Bax inhibition decreased cytosolic Smac and Cyto c release along with caspase activation and abrogated apoptosis, indicating that Smac and Bax are critical mediators of SapC-DOPS action. Similarly, pretreatment with the mitochondria-stabilizing agent bongkrekic acid decreased apoptosis indicating that loss of ΔΨM is critical for SapC-DOPS activity. Apoptosis induction was not critically dependent on reactive oxygen species (ROS) production and Cyclophilin D, since pretreatment with N-acetyl cysteine and cyclosporine A, respectively, did not prevent Smac or Cyto c release. CONCLUSIONS Taken together, our results indicate that SapC-DOPS acts through a mitochondria-mediated pathway accompanied by an early release of Smac and Bax. Specific tumor-targeting capacity and anticancer efficacy of SapC-DOPS supports its potential as a dual imaging and therapeutic agent in neuroblastoma therapy.
Collapse
Affiliation(s)
- Mahaboob K Sulaiman
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Zhengtao Chu
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
- Divison of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| | - Victor M Blanco
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Subrahmanya D Vallabhapurapu
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Robert S Franco
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Xiaoyang Qi
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
- Divison of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| |
Collapse
|
18
|
Costa LG, de Laat R, Tagliaferri S, Pellacani C. A mechanistic view of polybrominated diphenyl ether (PBDE) developmental neurotoxicity. Toxicol Lett 2014; 230:282-94. [PMID: 24270005 PMCID: PMC4028440 DOI: 10.1016/j.toxlet.2013.11.011] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/12/2013] [Indexed: 01/01/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs), extensively used in the past few decades as flame retardants in a variety of consumer products, have become world-wide persistent environmental pollutants. Levels in North America are usually higher than those in Europe and Asia, and body burden is 3-to-9-fold higher in infants and toddlers than in adults. The latter has raised concern for potential developmental toxicity and neurotoxicity of PBDEs. Experimental studies in animals and epidemiological observations in humans suggest that PBDEs may be developmental neurotoxicants. Pre- and/or post-natal exposure to PBDEs may cause long-lasting behavioral abnormalities, particularly in the domains of motor activity and cognition. The mechanisms underlying the developmental neurotoxic effects of PBDEs are not known, though several hypotheses have been put forward. One general mode of action relates to the ability of PBDEs to impair thyroid hormone homeostasis, thus indirectly affecting the developing brain. An alternative or additional mode of action involves a direct effect of PBDEs on nervous system cells; PBDEs can cause oxidative stress-related damage (DNA damage, mitochondrial dysfunction, apoptosis), and interfere with signal transduction (particularly calcium signaling), and with neurotransmitter systems. Important issues such as bioavailability and metabolism of PBDEs, extrapolation of results to low level of exposures, and the potential effects of interactions among PBDE congeners and between PBDEs and other contaminants also need to be taken into account.
Collapse
Affiliation(s)
- Lucio G Costa
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Department of Neuroscience, University of Parma, Parma, Italy.
| | - Rian de Laat
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
19
|
Westerink RHS. Modulation of cell viability, oxidative stress, calcium homeostasis, and voltage- and ligand-gated ion channels as common mechanisms of action of (mixtures of) non-dioxin-like polychlorinated biphenyls and polybrominated diphenyl ethers. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2014; 21:6373-6383. [PMID: 23686757 DOI: 10.1007/s11356-013-1759-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 04/22/2013] [Indexed: 06/02/2023]
Abstract
Non-dioxin-like polychlorinated biphenyls (NDL-PCBs) and polybrominated diphenyl ethers (PBDEs) are environmental pollutants that exert neurodevelopmental and neurobehavioral effects in vivo in humans and animals. Acute in vitro neurotoxic effects include changes in cell viability, oxidative stress, and basal intracellular calcium levels. Though these acute cellular effects could partly explain the observed in vivo effects, other mechanisms, such as effects on calcium influx and neurotransmitter receptor function, likely contribute to the disturbance in neurotransmission. This concise review combines in vitro data on cell viability, oxidative stress and basal calcium levels with recent data that clearly demonstrate that (hydroxylated) PCBs and (hydroxylated) PBDEs can exert acute effects on voltage-gated Ca(2+) channels as well as on excitatory and inhibitory neurotransmitter receptors in vitro. These novel mechanisms of action are shared by NDL-PCBs, OH-PBDEs, and some other persistent organic pollutants, such as tetrabromobisphenol-A, and could have profound effects on neurodevelopment, neurotransmission, and neurobehavior in vivo.
Collapse
Affiliation(s)
- Remco H S Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, 3508 TD, Utrecht, The Netherlands,
| |
Collapse
|
20
|
Asnake S, Pradhan A, Banjop-Kharlyngdoh J, Modig C, Olsson PE. 1,2-Dibromo-4-(1,2 dibromoethyl) cyclohexane (TBECH)-mediated steroid hormone receptor activation and gene regulation in chicken LMH cells. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2014; 33:891-899. [PMID: 24375616 DOI: 10.1002/etc.2509] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/28/2013] [Accepted: 12/16/2013] [Indexed: 06/03/2023]
Abstract
The incorporation of brominated flame retardants into industrial and household appliances has increased their occurrence in the environment, resulting in deleterious effects on wildlife. With the increasing restraints on available compounds, there has been a shift to using brominated flame retardants that has seen the production of alternative brominated flame retardants such as 1,2-dibromo-4-(1,2 dibromoethyl) cyclohexane (TBECH), which has been detected in the environment. In previous in silico and in vitro studies the authors have shown that TBECH can activate both the human androgen receptor (hAR) and the zebrafish AR (zAR) suggesting that it is a potential endocrine disruptor. The present study was aimed at determining the interaction of TBECH with the chicken AR (cAR). In the present study, TBECH bound to cAR, but in vitro activation assay studies using the chicken LMH cell line showed it had a potency of only 15% compared with testosterone. Sequence difference between ARs from different species may contribute to the different responses to TBECH. Further quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) analysis showed that TBECH interacted with and altered the expression of both thyroid receptors and estrogen receptors. In addition, the qRT-PCR analysis showed that TBECH altered the transcription pattern of genes involved in inflammatory, apoptotic, proliferative, DNA methylation, and drug-metabolizing pathways. This demonstrates that TBECH, apart from activating cAR, can also influence multiple biological pathways in the chicken.
Collapse
Affiliation(s)
- Solomon Asnake
- Örebro Life Science Center, Academy of Science, Technology, Örebro University, Örebro, Sweden
| | | | | | | | | |
Collapse
|
21
|
BDE-47 and 6-OH-BDE-47 modulate calcium homeostasis in primary fetal human neural progenitor cells via ryanodine receptor-independent mechanisms. Arch Toxicol 2014; 88:1537-48. [PMID: 24599297 DOI: 10.1007/s00204-014-1217-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 02/19/2014] [Indexed: 02/04/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are bioaccumulating flame retardants found in rising concentrations in human tissue. Epidemiological and animal studies have raised concern for their potential to induce developmental neurotoxicity (DNT). Considering the essential role of calcium homeostasis in neurodevelopment, PBDE-induced disturbance of intracellular calcium concentration ([Ca(2+)]i) may underlie PBDE-induced DNT. To test this hypothesis, we investigated acute effects of BDE-47 and 6-OH-BDE-47 on [Ca(2+)]i in human neural progenitor cells (hNPCs) and unraveled involved signaling pathways. Short-time differentiated hNPCs were exposed to BDE-47, 6-OH-BDE-47, and multiple inhibitors/stimulators of presumably involved signaling pathways to determine possible effects on [Ca(2+)]i by single-cell microscopy with the fluorescent dye Fura-2. Initial characterization of calcium signaling pathways confirmed the early developmental stage of hNPCs. In these cells, BDE-47 (2 μM) and 6-OH-BDE-47 (0.2 μM) induce [Ca(2+)]i transients. This increase in [Ca(2+)]i is due to extracellular Ca(2+) influx and intracellular release of Ca(2+), mainly from the endoplasmic reticulum (ER). While extracellular Ca(2+) seems to enter the cytoplasm upon 6-OH-BDE-47 by interfering with the cell membrane and independent of Ca(2+) ion channels, ER-derived Ca(2+) is released following activation of protein lipase C and inositol 1,4,5-trisphosphate receptor, but independently of ryanodine receptors. These findings illustrate that immature developing hNPCs respond to low concentrations of 6-OH-BDE-47 by an increase in [Ca(2+)]i and provide new mechanistic explanations for such BDE-induced calcium disruption. Thus, these data support the possibility of a critical window of PBDE exposure, i.e., early human brain development, which has to be acknowledged in risk assessment.
Collapse
|
22
|
Zhang S, Kuang G, Zhao G, Wu X, Zhang C, Lei R, Xia T, Chen J, Wang Z, Ma R, Li B, Yang L, Wang A. Involvement of the mitochondrial p53 pathway in PBDE-47-induced SH-SY5Y cells apoptosis and its underlying activation mechanism. Food Chem Toxicol 2013; 62:699-706. [DOI: 10.1016/j.fct.2013.10.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 09/15/2013] [Accepted: 10/05/2013] [Indexed: 12/31/2022]
|
23
|
Guo Y, Zhou B. Thyroid endocrine system disruption by pentachlorophenol: an in vitro and in vivo assay. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2013; 142-143:138-145. [PMID: 24001430 DOI: 10.1016/j.aquatox.2013.08.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/02/2013] [Accepted: 08/12/2013] [Indexed: 06/02/2023]
Abstract
The present study aimed to evaluate the disruption caused to the thyroid endocrine system by pentachlorophenol (PCP) using in vitro and in vivo assays. In the in vitro assay, rat pituitary GH3 cells were exposed to 0, 0.1, 0.3, and 1.0 μM PCP. PCP exposure significantly downregulated basal and triiodothyronine (T3)-induced Dio 1 transcription, indicating the antagonistic activity of PCP in vitro. In the in vivo assay, zebrafish embryos were exposed to 0, 1, 3, and 10 μg/L of PCP until 14 days post-fertilization. PCP exposure resulted in decreased thyroxine (T4) levels, but elevated contents of whole-body T3. PCP exposure significantly upregulated the mRNA expression of genes along hypothalamic-pituitary-thyroid (HPT) axis, including those encoding thyroid-stimulating hormone, sodium/iodide symporter, thyroglobulin, Dio 1 and Dio 2, alpha and beta thyroid hormone receptor, and uridinediphosphate-glucuronosyl-transferase. PCP exposure did not influence the transcription of the transthyretin (TTR) gene. The results indicate that PCP potentially disrupts the thyroid endocrine system both in vitro and in vivo.
Collapse
Affiliation(s)
- Yongyong Guo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | | |
Collapse
|
24
|
Bradner JM, Suragh TA, Wilson WW, Lazo CR, Stout KA, Kim HM, Wang MZ, Walker DI, Pennell KD, Richardson JR, Miller GW, Caudle WM. Exposure to the polybrominated diphenyl ether mixture DE-71 damages the nigrostriatal dopamine system: role of dopamine handling in neurotoxicity. Exp Neurol 2012; 241:138-47. [PMID: 23287494 DOI: 10.1016/j.expneurol.2012.12.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/13/2012] [Accepted: 12/18/2012] [Indexed: 11/26/2022]
Abstract
In the last several decades polybrominated diphenyl ethers (PBDEs) have replaced the previously banned polychlorinated biphenyls (PCBs) in multiple flame retardant utilities. As epidemiological and laboratory studies have suggested PCBs as a risk factor for Parkinson's disease (PD), the similarities between PBDEs and PCBs suggest that PBDEs have the potential to be neurotoxic to the dopamine system. The purpose of this study was to evaluate the neurotoxic effects of the PBDE mixture, DE-71, on the nigrostriatal dopamine system and address the role of altered dopamine handling in mediating this neurotoxicity. Using an in vitro model system we found DE-71 effectively caused cell death in a dopaminergic cell line as well as reducing the number of TH+ neurons isolated from VMAT2 WT and LO animals. Assessment of DE-71 neurotoxicity in vivo demonstrated significant deposition of PBDE congeners in the brains of mice, leading to reductions in striatal dopamine and dopamine handling, as well as reductions in the striatal dopamine transporter (DAT) and VMAT2. Additionally, DE-71 elicited a significant locomotor deficit in the VMAT2 WT and LO mice. However, no change was seen in TH expression in dopamine terminal or in the number of dopamine neurons in the substantia nigra pars compacta (SNpc). To date, these are the first data to demonstrate that exposure to PBDEs disrupts the nigrostriatal dopamine system. Given their similarities to PCBs, additional laboratory and epidemiological research should be considered to assess PBDEs as a potential risk factor for PD and other neurological disorders.
Collapse
Affiliation(s)
- Joshua M Bradner
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322-3090, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Schwartzer JJ, Koenig CM, Berman RF. Using mouse models of autism spectrum disorders to study the neurotoxicology of gene-environment interactions. Neurotoxicol Teratol 2012; 36:17-35. [PMID: 23010509 DOI: 10.1016/j.ntt.2012.08.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 08/28/2012] [Accepted: 08/30/2012] [Indexed: 10/27/2022]
Abstract
To better study the role of genetics in autism, mouse models have been developed which mimic the genetics of specific autism spectrum and related disorders. These models have facilitated research on the role genetic susceptibility factors in the pathogenesis of autism in the absence of environmental factors. Inbred mouse strains have been similarly studied to assess the role of environmental agents on neurodevelopment, typically without the complications of genetic heterogeneity of the human population. What has not been as actively pursued, however, is the methodical study of the interaction between these factors (e.g., gene and environmental interactions in neurodevelopment). This review suggests that a genetic predisposition paired with exposure to environmental toxicants plays an important role in the etiology of neurodevelopmental disorders including autism, and may contribute to the largely unexplained rise in the number of children diagnosed with autism worldwide. Specifically, descriptions of the major mouse models of autism and toxic mechanisms of prevalent environmental chemicals are provided followed by a discussion of current and future research strategies to evaluate the role of gene and environment interactions in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jared J Schwartzer
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Davis, Davis, CA 95618, United States.
| | | | | |
Collapse
|
26
|
Jiang C, Zhang S, Liu H, Zeng Q, Xia T, Chen Y, Kuang G, Zhao G, Wu X, Zhang X, Wang A. The role of the IRE1 pathway in PBDE-47-induced toxicity in human neuroblastoma SH-SY5Y cells in vitro. Toxicol Lett 2012; 211:325-33. [DOI: 10.1016/j.toxlet.2012.04.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 04/05/2012] [Accepted: 04/12/2012] [Indexed: 10/28/2022]
|
27
|
Dingemans MML, van den Berg M, Westerink RHS. Neurotoxicity of brominated flame retardants: (in)direct effects of parent and hydroxylated polybrominated diphenyl ethers on the (developing) nervous system. ENVIRONMENTAL HEALTH PERSPECTIVES 2011; 119:900-7. [PMID: 21245014 PMCID: PMC3223008 DOI: 10.1289/ehp.1003035] [Citation(s) in RCA: 243] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Accepted: 01/18/2011] [Indexed: 05/17/2023]
Abstract
BACKGROUND/OBJECTIVE Polybrominated diphenyl ethers (PBDEs) and their hydroxylated (OH-) or methoxylated forms have been detected in humans. Because this raises concern about adverse effects on the developing brain, we reviewed the scientific literature on these mechanisms. DATA SYNTHESIS Many rodent studies reported behavioral changes after developmental, neonatal, or adult exposure to PBDEs, and other studies documented subtle structural and functional alterations in brains of PBDE-exposed animals. Functional effects have been observed on synaptic plasticity and the glutamate-nitric oxide-cyclic guanosine monophosphate pathway. In the brain, changes have been observed in the expression of genes and proteins involved in synapse and axon formation, neuronal morphology, cell migration, synaptic plasticity, ion channels, and vesicular neurotransmitter release. Cellular and molecular mechanisms include effects on neuronal viability
(via apoptosis and oxidative stress), neuronal differentiation and migration, neurotransmitter release/uptake, neurotransmitter receptors and ion channels, calcium (Ca²⁺) homeostasis, and intracellular signaling pathways. DISCUSSION Bioactivation of PBDEs by hydroxylation has been observed for several endocrine end points. This has also been observed for mechanisms related to neurodevelopment, including binding to thyroid hormone receptors and transport proteins, disruption of Ca²⁺ homeostasis, and modulation of GABA and nicotinic acetylcholine receptor function. CONCLUSIONS The increased hazard for developmental neurotoxicity by hydroxylated (OH-)PBDEs compared with their parent congeners via direct neurotoxicity and thyroid disruption clearly warrants further investigation into a) the role of oxidative metabolism in producing active metabolites of PBDEs and their impact on brain development; b) concentrations of parent and OH-PBDEs in the brain; and c) interactions between different environmental contaminants during exposure to mixtures, which may increase neurotoxicity.
Collapse
Affiliation(s)
- Milou M L Dingemans
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands.
| | | | | |
Collapse
|
28
|
Verner MA, Bouchard M, Fritsche E, Charbonneau M, Haddad S. In vitro neurotoxicity data in human risk assessment of polybrominated diphenyl ethers (PBDEs): overview and perspectives. Toxicol In Vitro 2011; 25:1509-15. [PMID: 21704695 DOI: 10.1016/j.tiv.2011.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 04/20/2011] [Accepted: 06/10/2011] [Indexed: 12/30/2022]
Abstract
Polybrominated diphenyl ethers (PBDEs) are flame retardants routinely detected in samples of cord blood and breast milk. Concerns have been raised with regard to the toxicity of both pre- and postnatal exposures towards the developing nervous system. Although there is an increasing body of literature on the disruption of brain cell functions by certain PBDE congeners in vitro, some challenges have yet to be tackled to enable the translation of in vitro findings into their in vivo counterparts. In this paper, we review findings on the PBDE neurotoxicity in human cells and discuss the research gaps to be addressed. Moreover, we propose a scheme for the incorporation of in vitro data in human risk assessment, namely through (i) the determination of in vitro cell benchmark levels; (ii) the consideration of uncertainties in establishing equivalency between the in vitro and the in vivo tissue benchmark levels (e.g., chronic vs. acute exposure, interactions with other chemicals); and (iii) relating tissue benchmark levels to surrogate levels of internal exposure. Alongside the assessment of brain dosimetry following exposure to PBDEs, in vitro neurotoxicity data provide a unique opportunity to evaluate the risks of prenatal and early life exposures on children neurodevelopment.
Collapse
Affiliation(s)
- Marc-André Verner
- TOXEN, Département des Sciences Biologiques, Université du Québec à Montréal, C.P. 8888 Succ. Centre-Ville, Montreal, Canada H3C 3P8.
| | | | | | | | | |
Collapse
|
29
|
Zanchetta LM, Garcia A, Lyng F, Walsh J, Murphy JEJ. Mitophagy and mitochondrial morphology in human melanoma-derived cells post exposure to simulated sunlight. Int J Radiat Biol 2011; 87:506-17. [PMID: 21381890 DOI: 10.3109/09553002.2011.556175] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE To assess changes in mitochondrial morphology and mitophagy induced by simulated sunlight irradiation (SSI) and how these changes are modulated by mitochondrial activity and energy source. MATERIALS AND METHODS Human malignant amelanotic melanoma A375 cells were pre-treated with either a mitochondrial activity enhancer, uncoupler or were either melanin or glutamine supplemented/starved for 4 hours pre-exposure to sunlight. A Q-Sun Solar Simulator (Q-Lab, Homestead, FL, USA) was employed to expose cells to simulated sunlight. Confocal microscopy imaging of A375 cells co-loaded with mitochondria and lysosome-specific fluorescent dyes was used to identify these organelles and predict mitophagic events. RESULTS SSI induces pronounced changes in mitochondrial dynamics and mitophagy in exposed skin cells compared to control and these effects were modified by both glutamine and melanin. CONCLUSIONS Mitochondrial dynamics and rate of mitophagy in melanoma cells are sensitive to even short bursts of environmentally relevant SSI. Mitochondrial dynamics, and its modulation, may also play a role in mitophagy regulation, cell survival and proliferation post SSI.
Collapse
Affiliation(s)
- Luciene M Zanchetta
- Mitochondrial Biology & Radiation Research, School of Science, Institute of Technology Sligo, Ash Lane, Sligo, Ireland.
| | | | | | | | | |
Collapse
|
30
|
|
31
|
Wang D, Li QX. Application of mass spectrometry in the analysis of polybrominated diphenyl ethers. MASS SPECTROMETRY REVIEWS 2010; 29:737-775. [PMID: 19722247 DOI: 10.1002/mas.20263] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
This review summarized the applications of mass spectrometric techniques for the analysis of the important flame retardants polybrominated diphenyl ethers (PBDEs) to understand the environmental sources, fate and toxicity of PBDEs that were briefly discussed to give a general idea for the need of analytical methodologies. Specific performance of various mass spectrometers hyphenated with, for example, gas chromatograph, liquid chromatograph, and inductively coupled plasma (GC/MS, LC/MS, and ICP/MS, respectively) for the analysis of PBDEs was compared with an objective to present the information on the evolution of MS techniques for determining PBDEs in environmental and human samples. GC/electron capture negative ionization quadrupole MS (GC/NCI qMS), GC/high resolution MS (GC/HRMS) and GC ion trap MS (GC/ITMS) are most commonly used MS techniques for the determination of PBDEs. New analytical technologies such as fast tandem GC/MS and LC/MS become available to improve analyses of higher PBDEs. The development and application of the tandem MS techniques have helped to understand environmental fate and transformations of PBDEs of which abiotic and biotic degradation of decaBDE is thought to be one major source of Br(1-9)BDEs present in the environment in addition to direct loading from commercial mixtures. MS-based proteomics will offer an insight into the molecular mechanisms of toxicity and potential developmental and neurotoxicity of PBDEs.
Collapse
Affiliation(s)
- Dongli Wang
- Department of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, Hawaii 96822, USA
| | | |
Collapse
|
32
|
Jin S, Yang F, Hui Y, Xu Y, Lu Y, Liu J. Cytotoxicity and apoptosis induction on RTG-2 cells of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) and decabrominated diphenyl ether (BDE-209). Toxicol In Vitro 2010; 24:1190-6. [PMID: 20159034 DOI: 10.1016/j.tiv.2010.02.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 12/27/2009] [Accepted: 02/09/2010] [Indexed: 10/19/2022]
Abstract
Recently, the environmental residues of polybrominated diphenyl ethers (PBDEs) have markedly increased. In particular, the levels of certain PBDE congeners in fish have raised concern regarding potential risks associated with dietary PBDEs exposures. However, little is known regarding PBDE-mediated cell injury in relevant in vitro fish cell models. In this study, the cytotoxic effects of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) and decabrominated diphenyl ether (BDE-209) on RTG-2 cells were investigated. RTG-2 cells were incubated with different concentrations of BDE-47 and BDE-209 (1-100 microM) for 72 h, and a set of bioassays were conducted to measure: cell viability (evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and neutral red (NR) uptake), lactate dehydrogenase (LDH) leakage, reactive oxygen species (ROS) formation and cell apoptosis. The results showed that BDE-47 and BDE-209 inhibited the cells viability, increased LDH leakage, and induced cell apoptosis in time and concentration-dependent manner. All significant effects were observed at concentrations of 12.5 microM and above for BDE-47 and 25 microM and above for BDE-209 (P<0.05). At the concentration of 100 microM BDE-47 and BDE-209, the cell viability of the exposed cells dropped to about 40% and 50% of the control, and the apoptotic rates were 52.6% and 34.6%, respectively. After 12h exposure, a concentration-dependent increases of BDE-47 and BDE-209 (12.5-100 microM) in ROS formation were observed. Collectively, the results of cell viability, LDH leakage, cell apoptosis and ROS formation demonstrated that the toxic mechanism of PBDEs on RTG-2 might be mediated by oxidative stress.
Collapse
Affiliation(s)
- Shiwei Jin
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430073, China
| | | | | | | | | | | |
Collapse
|
33
|
Alm H, Scholz B, Kultima K, Nilsson A, Andrén PE, Savitski MM, Bergman Å, Stigson M, Fex-Svenningsen Å, Dencker L. In Vitro Neurotoxicity of PBDE-99: Immediate and Concentration-Dependent Effects on Protein Expression in Cerebral Cortex Cells. J Proteome Res 2009; 9:1226-35. [DOI: 10.1021/pr900723c] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Henrik Alm
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Birger Scholz
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Kim Kultima
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Per E. Andrén
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Mikhail M. Savitski
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Åke Bergman
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Michael Stigson
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Åsa Fex-Svenningsen
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| | - Lennart Dencker
- Department of Pharmaceutical Biosciences, Division of Toxicology, Uppsala University, Sweden, Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Sweden, Department of Cellular and Molecular Biology, Uppsala University, Sweden, Department of Environmental Chemistry, Stockholm University, Sweden, and Institute of Medical Biology, Anatomy and Neurobiology, University of Southern Denmark, Denmark
| |
Collapse
|
34
|
Fonnum F, Mariussen E. Mechanisms involved in the neurotoxic effects of environmental toxicants such as polychlorinated biphenyls and brominated flame retardants. J Neurochem 2009; 111:1327-47. [DOI: 10.1111/j.1471-4159.2009.06427.x] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|